Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 221(2): 313-324, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31250000

RESUMEN

BACKGROUND: During pregnancy, the Zika flavivirus (ZIKV) infects human placentas, inducing defects in the developing fetus. The flavivirus nonstructural protein 1 (NS1) alters glycosaminoglycans on the endothelium, causing hyperpermeability in vitro and vascular leakage in vivo in a tissue-dependent manner. The contribution of ZIKV NS1 to placental dysfunction during ZIKV infection remains unknown. METHODS: We examined the effect of ZIKV NS1 on expression and release of heparan sulfate (HS), hyaluronic acid (HA), and sialic acid on human trophoblast cell lines and anchoring villous explants from first-trimester placentas infected with ZIKV ex vivo. We measured changes in permeability in trophoblasts and stromal cores using a dextran-based fluorescence assay and changes in HA receptor expression using immunofluorescent microscopy. RESULTS: ZIKV NS1 in the presence and absence of ZIKV increased the permeability of anchoring villous explants. ZIKV NS1 induced shedding of HA and HS and altered expression of CD44 and lymphatic endothelial cell HA receptor-1, HA receptors on stromal fibroblasts and Hofbauer macrophages in villous cores. Hyaluronidase was also stimulated in NS1-treated trophoblasts. CONCLUSIONS: These findings suggest that ZIKV NS1 contributes to placental dysfunction via modulation of glycosaminoglycans on trophoblasts and chorionic villi, resulting in increased permeability of human placentas.


Asunto(s)
Placenta/metabolismo , Proteínas no Estructurales Virales/metabolismo , Infección por el Virus Zika/transmisión , Virus Zika/metabolismo , Femenino , Glicosaminoglicanos/metabolismo , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Permeabilidad , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/virología , Infección por el Virus Zika/virología
2.
Med Microbiol Immunol ; 208(3-4): 475-485, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31065796

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, yet there are no established treatments for preventing maternal-fetal transmission. During first trimester, HCMV replicates in basal decidua that functions as a reservoir for virus and source of transmission to the attached placenta and fetal hemiallograft but also contains immune cells, including natural killer cells, macrophages, and T cell subsets, that respond to pathogens, protecting the placenta and fetus. However, the specific cellular and cytokine responses to infection are unknown, nor are the immune correlates of protection that guide development of therapeutic strategies. Here we survey immune cell phenotypes in intact explants of basal decidua infected with a clinical pathogenic HCMV strain ex vivo and identify specific changes occurring in response to infection in the tissue environment. Using 4-color immunofluorescence microscopy, we found that at 3 days postinfection, virus replicates in decidual stromal cells and epithelial cells of endometrial glands. Infected cells and effector memory CD8+ T cells (TEM) in contact with them make IFN-γ. CD8+ TEM cells produce granulysin and cluster at sites of infection in decidua and the epithelium of endometrial glands. Quantification indicated expansion of two immune cell subtypes-CD8+ TEM cells and, to a lesser extent, iNKT cells. Approximately 20% of immune cells were found in pairs in both control and infected decidua, suggesting frequent cross-talk in the microenvironment of decidua. Our findings indicate a complex immune microenvironment in basal decidua and suggest CD8+ TEM cells play a role in early responses to decidual infection in seropositive women.


Asunto(s)
Infecciones por Citomegalovirus/patología , Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/inmunología , Decidua/patología , Inmunidad Celular , Placenta/patología , Linfocitos T CD8-positivos/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Humanos , Células T Asesinas Naturales/inmunología , Técnicas de Cultivo de Órganos , Embarazo , Células del Estroma/patología , Células del Estroma/virología
3.
J Infect Dis ; 217(8): 1202-1213, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29106643

RESUMEN

Background: Maternal Zika virus (ZIKV) infection with prolonged viremia leads to fetal infection and congenital Zika syndrome. Previously, we reported that ZIKV infects primary cells from human placentas and fetal membranes. Here, we studied viral replication in numerous explants of anchoring villi and basal decidua from first-trimester human placentas and midgestation amniotic epithelial cells (AmEpCs). Methods: Explants and AmEpCs were infected with American and African ZIKV strains at low multiplicities, and ZIKV proteins were visualized by immunofluorescence. Titers of infectious progeny, cell proliferation, and invasiveness were quantified. Results: In anchoring villus, ZIKV replicated reproducibly in proliferating cytotrophoblasts in proximal cell columns, dividing Hofbauer cells in villus cores, and invasive cytotrophoblasts, but frequencies differed. Cytotrophoblasts in explants infected by Nicaraguan strains were invasive, whereas those infected by prototype MR766 largely remained in cell columns, and titers varied by donor and strain. In basal decidua, ZIKV replicated in glandular epithelium, decidual cells, and immune cells. ZIKV-infected AmEpCs frequently occurred in pairs and expressed Ki67 and phosphohistone H3, indicating replication in dividing cells. Conclusions: ZIKV infection in early pregnancy could target proliferating cell column cytotrophoblasts and Hofbauer cells, amplifying infection in basal decidua and chorionic villi and enabling transplacental transmission.


Asunto(s)
Complicaciones Infecciosas del Embarazo/virología , Replicación Viral/fisiología , Infección por el Virus Zika/virología , Virus Zika/química , Amnios/citología , Células Epiteliales/virología , Femenino , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Placenta/virología , Embarazo , Primer Trimestre del Embarazo , Virus Zika/genética
4.
Am J Pathol ; 186(11): 2970-2986, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27638253

RESUMEN

Human cytomegalovirus (HCMV) is the leading viral cause of birth defects, including microcephaly, neurological deficits, hearing impairment, and vision loss. We previously reported that epithelial cells in amniotic membranes of placentas from newborns with intrauterine growth restriction and underlying congenital HCMV infection contain viral proteins in cytoplasmic vesicles. Herein, we immunostained amniotic membranes from 51 placentas from symptomatic and asymptomatic congenital infection with HCMV DNA in amniotic fluid and/or newborn saliva, intrauterine growth restriction, preterm deliveries, and controls. We consistently observed HCMV proteins in amniotic epithelial cells (AmEpCs) from infected placentas, sometimes with aberrant morphology. Primary AmEpCs isolated from mid-gestation placentas infected with pathogenic VR1814 proliferated and released infectious progeny for weeks, producing higher virus titers than late-gestation cells that varied by donor. In contrast to intact virion assembly compartments in differentiated retinal pigment epithelial cells, infected AmEpCs made dispersed multivesicular bodies. Primary AmEpCs and explants of amniochorionic membranes from mid-gestation placentas formed foci of infection, and interferon-ß production was prolonged. Infected AmEpCs up-regulated anti-apoptotic proteins survivin and Bcl-xL by mechanisms dependent and independent of the activated STAT3. Amniotic membranes naturally expressed both survivin and Bcl-xL, indicating that fetal membranes could foster persistent viral infection. Our results suggest strengthening innate immune responses and reducing viral functions could suppress HCMV infection in the fetal compartment.


Asunto(s)
Infecciones por Citomegalovirus/congénito , Citomegalovirus/inmunología , Placenta/virología , Complicaciones Infecciosas del Embarazo/virología , Amnios/patología , Amnios/virología , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Femenino , Retardo del Crecimiento Fetal/virología , Feto/metabolismo , Edad Gestacional , Humanos , Recién Nacido , Interferón beta/metabolismo , Placenta/patología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/patología , Carga Viral , Replicación Viral
5.
J Virol ; 89(9): 5134-47, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25741001

RESUMEN

UNLABELLED: Human cytomegalovirus (HCMV) is a major cause of birth defects that include severe neurological deficits, hearing and vision loss, and intrauterine growth restriction. Viral infection of the placenta leads to development of avascular villi, edema, and hypoxia associated with symptomatic congenital infection. Studies of primary cytotrophoblasts (CTBs) revealed that HCMV infection impedes terminal stages of differentiation and invasion by various molecular mechanisms. We recently discovered that HCMV arrests earlier stages involving development of human trophoblast progenitor cells (TBPCs), which give rise to the mature cell types of chorionic villi-syncytiotrophoblasts on the surfaces of floating villi and invasive CTBs that remodel the uterine vasculature. Here, we show that viral proteins are present in TBPCs of the chorion in cases of symptomatic congenital infection. In vitro studies revealed that HCMV replicates in continuously self-renewing TBPC lines derived from the chorion and alters expression and subcellular localization of proteins required for cell cycle progression, pluripotency, and early differentiation. In addition, treatment with a human monoclonal antibody to HCMV glycoprotein B rescues differentiation capacity, and thus, TBPCs have potential utility for evaluation of the efficacies of novel antiviral antibodies in protecting and restoring placental development. Our results suggest that HCMV replicates in TBPCs in the chorion in vivo, interfering with the earliest steps in the growth of new villi, contributing to virus transmission and impairing compensatory development. In cases of congenital infection, reduced responsiveness of the placenta to hypoxia limits the transport of substances from maternal blood and contributes to fetal growth restriction. IMPORTANCE: Human cytomegalovirus (HCMV) is a leading cause of birth defects in the United States. Congenital infection can result in permanent neurological defects, mental retardation, hearing loss, visual impairment, and pregnancy complications, including intrauterine growth restriction, preterm delivery, and stillbirth. Currently, there is neither a vaccine nor any approved treatment for congenital HCMV infection during gestation. The molecular mechanisms underlying structural deficiencies in the placenta that undermine fetal development are poorly understood. Here we report that HCMV replicates in trophoblast progenitor cells (TBPCs)-precursors of the mature placental cells, syncytiotrophoblasts and cytotrophoblasts, in chorionic villi-in clinical cases of congenital infection. Virus replication in TBPCs in vitro dysregulates key proteins required for self-renewal and differentiation and inhibits normal division and development into mature placental cells. Our findings provide insights into the underlying molecular mechanisms by which HCMV replication interferes with placental maturation and transport functions.


Asunto(s)
Diferenciación Celular , Infecciones por Citomegalovirus/patología , Citomegalovirus/fisiología , Placenta/virología , Células Madre/virología , Trofoblastos/virología , Replicación Viral , Infecciones por Citomegalovirus/virología , Femenino , Humanos , Embarazo , Complicaciones Infecciosas del Embarazo/patología , Complicaciones Infecciosas del Embarazo/virología , Células Madre/fisiología , Trofoblastos/fisiología
6.
Antimicrob Agents Chemother ; 59(3): 1558-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534746

RESUMEN

Human cytomegalovirus (HCMV) is the most common infection causing poor outcomes among transplant recipients. Maternal infection and transplacental transmission are major causes of permanent birth defects. Although no active vaccines to prevent HCMV infection have been approved, passive immunization with HCMV-specific immunoglobulin has shown promise in the treatment of both transplant and congenital indications. Antibodies targeting the viral glycoprotein B (gB) surface protein are known to neutralize HCMV infectivity, with high-affinity binding being a desirable trait, both to compete with low-affinity antibodies that promote the transmission of virus across the placenta and to displace nonneutralizing antibodies binding nearby epitopes. Using a miniaturized screening technology to characterize secreted IgG from single human B lymphocytes, 30 antibodies directed against gB were previously cloned. The most potent clone, TRL345, is described here. Its measured affinity was 1 pM for the highly conserved site I of the AD-2 epitope of gB. Strain-independent neutralization was confirmed for 15 primary HCMV clinical isolates. TRL345 prevented HCMV infection of placental fibroblasts, smooth muscle cells, endothelial cells, and epithelial cells, and it inhibited postinfection HCMV spread in epithelial cells. The potential utility for preventing congenital transmission is supported by the blockage of HCMV infection of placental cell types central to virus transmission to the fetus, including differentiating cytotrophoblasts, trophoblast progenitor cells, and placental fibroblasts. Further, TRL345 was effective at controlling an ex vivo infection of human placental anchoring villi. TRL345 has been utilized on a commercial scale and is a candidate for clinical evaluation.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Linfocitos B/virología , Línea Celular , Infecciones por Citomegalovirus/virología , Células Endoteliales/inmunología , Células Endoteliales/virología , Células Epiteliales/inmunología , Células Epiteliales/virología , Epítopos/inmunología , Femenino , Fibroblastos/inmunología , Fibroblastos/virología , Humanos , Inmunoglobulina G/inmunología , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/virología , Placenta/inmunología , Placenta/virología , Embarazo , Proteínas del Envoltorio Viral/inmunología
7.
J Infect Dis ; 209(10): 1573-84, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24403553

RESUMEN

BACKGROUND: Human cytomegalovirus (HCMV) is the major viral etiology of congenital infection and birth defects. Fetal transmission is high (30%-40%) in primary maternal infection, and symptomatic babies have permanent neurological, hearing, and vision defects. Recurrent infection is infrequently transmitted (2%) and largely asymptomatic. Congenital infection is also associated with intrauterine growth restriction (IUGR). METHODS: To investigate possible underlying HCMV infection in cases of idiopathic IUGR, we studied maternal and cord sera and placentas from 19 pregnancies. Anti-HCMV antibodies, hypoxia-related factors, and cmvIL-10 were measured in sera. Placental biopsy specimens were examined for viral DNA, expression of infected cell proteins, and pathology. RESULTS: Among 7 IUGR cases, we identified 2 primary and 3 recurrent HCMV infections. Virus replicated in glandular epithelium and lymphatic endothelium in the decidua, cytotrophoblasts, and smooth muscle cells in blood vessels of floating villi and the chorion. Large fibrinoids with avascular villi, edema, and inflammation were significantly increased. Detection of viral proteins in the amniotic epithelium indicated transmission in 2 cases of IUGR with primary infection and 3 asymptomatic recurrent infections. CONCLUSIONS: Congenital HCMV infection impairs placental development and functions and should be considered as an underlying cause of IUGR, regardless of virus transmission to the fetus.


Asunto(s)
Infecciones por Citomegalovirus/complicaciones , Retardo del Crecimiento Fetal/virología , Complicaciones Infecciosas del Embarazo/patología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , ADN Viral , Femenino , Humanos , Inmunoglobulina G/sangre , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Proyectos Piloto , Embarazo , Pruebas Serológicas
8.
Clin Infect Dis ; 57 Suppl 4: S174-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24257421

RESUMEN

Human cytomegalovirus (HCMV) infection is transmitted from the infected mother to the placenta and fetus. Virus replicates in the decidua, invasive cytotrophoblasts that breach the uterine vasculature and villous cytotrophoblasts underlying syncytiotrophoblasts, then reaches blood vessels in the villus core. Virus replication, fibrosis, and edema result in a hypoxic intrauterine environment and release of cytokines that stimulates compensatory development of the placenta. We employed villous explant cultures to study viral effects on differentiation and test novel approaches to rescue the placenta from infection.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , Citomegalovirus/inmunología , Femenino , Humanos , Inmunoglobulinas/inmunología , Modelos Biológicos , Placenta/inmunología , Placenta/virología , Embarazo , Útero/inmunología , Útero/virología , Replicación Viral
9.
Am J Pathol ; 181(5): 1540-59, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22959908

RESUMEN

We investigated human cytomegalovirus pathogenesis by comparing infection with the low-passage, endotheliotropic strain VR1814 and the attenuated laboratory strain AD169 in human placental villi as explants in vitro and xenografts transplanted into kidney capsules of SCID mice (ie, mice with severe combined immunodeficiency). In this in vivo human placentation model, human cytotrophoblasts invade the renal parenchyma, remodel resident arteries, and induce a robust lymphangiogenic response. VR1814 replicated in villous and cell column cytotrophoblasts and reduced formation of anchoring villi in vitro. In xenografts, infected cytotrophoblasts had a severely diminished capacity to invade and remodel resident arteries. Infiltrating lymphatic endothelial cells proliferated, aggregated, and failed to form lymphatic vessels. In contrast, AD169 grew poorly in cytotrophoblasts in explants, and anchoring villi formed normally in vitro. Likewise, viral replication was impaired in xenografts, and cytotrophoblasts retained invasive capacity, but some partially remodeled blood vessels incorporated lymphatic endothelial cells and were permeable to blood. The expression of both vascular endothelial growth factor (VEGF)-C and basic fibroblast growth factor increased in VR1814-infected explants, whereas VEGF-A and soluble VEGF receptor-3 increased in those infected with AD169. Our results suggest that viral replication and paracrine factors could undermine vascular remodeling and cytotrophoblast-induced lymphangiogenesis, contributing to bleeding, hypoxia, and edema in pregnancies complicated by congenital human cytomegalovirus infection.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Infecciones por Citomegalovirus/patología , Citomegalovirus/fisiología , Linfangiogénesis , Placentación , Trofoblastos/patología , Trofoblastos/virología , Animales , Arterias/patología , Vasos Sanguíneos/patología , Movimiento Celular , Proliferación Celular , Vellosidades Coriónicas/crecimiento & desarrollo , Vellosidades Coriónicas/patología , Vellosidades Coriónicas/trasplante , Vellosidades Coriónicas/virología , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/fisiopatología , Infecciones por Citomegalovirus/virología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Riñón/irrigación sanguínea , Riñón/patología , Ratones , Ratones SCID , Embarazo , Células Madre/patología , Células Madre/virología , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Replicación Viral
10.
Vaccines (Basel) ; 10(7)2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35891239

RESUMEN

Human cytomegalovirus (HCMV) is the leading viral cause of congenital disease and permanent birth defects worldwide. Although the development of an effective vaccine is a public health priority, no vaccines are approved. Among the major antigenic targets are glycoproteins in the virion envelope, including gB, which facilitates cellular entry, and the pentameric complex (gH/gL/pUL128-131), required for the infection of specialized cell types. In this study, sera from rabbits immunized with the recombinant pentameric complex were tested for their ability to neutralize infection of epithelial cells, fibroblasts, and primary placental cell types. Sera from rhesus macaques immunized with recombinant gB or gB plus pentameric complex were tested for HCMV neutralizing activity on both cultured cells and cell column cytotrophoblasts in first-trimester chorionic villus explants. Sera from rabbits immunized with the pentameric complex potently blocked infection by pathogenic viral strains in amniotic epithelial cells and cytotrophoblasts but were less effective in fibroblasts and trophoblast progenitor cells. Sera from rhesus macaques immunized with the pentameric complex and gB more strongly reduced infection in fibroblasts, epithelial cells, and chorionic villus explants than sera from immunization with gB alone. These results suggest that the pentameric complex and gB together elicit antibodies that could have potential as prophylactic vaccine antigens.

11.
Am J Pathol ; 177(3): 1298-310, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20651234

RESUMEN

Human cytomegalovirus (HCMV) is the major viral cause of birth defects worldwide. Affected infants can have temporary symptoms that resolve soon after birth, such as growth restriction, and permanent disabilities, including neurological impairment. Passive immunization of pregnant women with primary HCMV infection is a promising treatment to prevent congenital disease. To understand the effects of sustained viral replication on the placenta and passive transfer of protective antibodies, we performed immunohistological analysis of placental specimens from women with untreated congenital infection, HCMV-specific hyperimmune globulin treatment, and uninfected controls. In untreated infection, viral replication proteins were found in trophoblasts and endothelial cells of chorionic villi and uterine arteries. Associated damage included extensive fibrinoid deposits, fibrosis, avascular villi, and edema, which could impair placental functions. Vascular endothelial growth factor and its receptor fms-like tyrosine kinase 1 (Flt1) were up-regulated, and amniotic fluid contained elevated levels of soluble Flt1 (sFlt1), an antiangiogenic protein, relative to placental growth factor. With hyperimmune globulin treatment, placentas appeared uninfected, vascular endothelial growth factor and Flt1 expression was reduced, and sFlt1 levels in amniotic fluid were lower. An increase in the number of chorionic villi and blood vessels over that in controls suggested compensatory development for a hypoxia-like condition. Taken together the results indicate that antibody treatment can suppress HCMV replication and prevent placental dysfunction, thus improving fetal outcome.


Asunto(s)
Vellosidades Coriónicas/virología , Infecciones por Citomegalovirus/metabolismo , Placenta/virología , Trofoblastos/virología , Proteínas Virales/metabolismo , Análisis de Varianza , Vellosidades Coriónicas/metabolismo , Vellosidades Coriónicas/patología , Citomegalovirus , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Placenta/metabolismo , Placenta/patología , Embarazo , Trofoblastos/metabolismo , Trofoblastos/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
Vaccines (Basel) ; 7(4)2019 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-31569508

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects worldwide, yet the most effective strategies for preventing virus transmission during pregnancy are unknown. We measured the efficacy of human monoclonal antibodies (mAbs) to HCMV attachment/entry factors glycoprotein B (gB) and the pentameric complex, gH/gL-pUL128-131, in preventing infection and spread of a clinical strain in primary placental cells and explants of developing anchoring villi. A total of 109 explants from five first-trimester placentas were cultured, and infection was analyzed in over 400 cell columns containing ~120,000 cytotrophoblasts (CTBs). mAbs to gB and gH/gL, 3-25 and 3-16, respectively, neutralized infection in stromal fibroblasts and trophoblast progenitor cells. mAbs to pUL128-131 of the pentameric complex, 1-103 and 2-18, neutralized infection of amniotic epithelial cells better than mAbs 3-25 and 3-16 and hyperimmune globulin. Select mAbs neutralized infection of cell column CTBs, with mAb 2-18 most effective, followed by mAb 3-25. Treatment of anchoring villi with mAbs postinfection reduced spread in CTBs and impaired formation of virion assembly compartments, with mAb 2-18 achieving better suppression at lower concentrations. These results predict that antibodies generated by HCMV vaccines or used for passive immunization have the potential to reduce transplacental transmission and congenital disease.

13.
Annu Rev Virol ; 5(1): 273-299, 2018 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-30048217

RESUMEN

Why certain viruses cross the physical barrier of the human placenta but others do not is incompletely understood. Over the past 20 years, we have gained deeper knowledge of intrauterine infection and routes of viral transmission. This review focuses on human viruses that replicate in the placenta, infect the fetus, and cause birth defects, including rubella virus, varicella-zoster virus, parvovirus B19, human cytomegalovirus (CMV), Zika virus (ZIKV), and hepatitis E virus type 1. Detailed discussions include ( a) the architecture of the uterine-placental interface, ( b) studies of placental explants ex vivo that provide insights into the infection and spread of CMV and ZIKV to the fetal compartment and how these viruses undermine early development, and ( c) novel treatments and vaccines that limit viral replication and have the potential to reduce dissemination, vertical transmission and the occurrence of congenital disease.


Asunto(s)
Transmisión Vertical de Enfermedad Infecciosa , Intercambio Materno-Fetal , Complicaciones Infecciosas del Embarazo/virología , Virosis/congénito , Femenino , Humanos , Placenta/virología , Embarazo
14.
Endocrinology ; 148(3): 1059-79, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17170095

RESUMEN

Human placentation entails the remarkable integration of fetal and maternal cells into a single functional unit. In the basal plate region (the maternal-fetal interface) of the placenta, fetal cytotrophoblasts from the placenta invade the uterus and remodel the resident vasculature and avoid maternal immune rejection. Knowing the molecular bases for these unique cell-cell interactions is important for understanding how this specialized region functions during normal pregnancy with implications for tumor biology and transplantation immunology. Therefore, we undertook a global analysis of the gene expression profiles at the maternal-fetal interface. Basal plate biopsy specimens were obtained from 36 placentas (14-40 wk) at the conclusion of normal pregnancies. RNA was isolated, processed, and hybridized to HG-U133A&B Affymetrix GeneChips. Surprisingly, there was little change in gene expression during the 14- to 24-wk interval. In contrast, 418 genes were differentially expressed at term (37-40 wk) as compared with midgestation (14-24 wk). Subsequent analyses using quantitative PCR and immunolocalization approaches validated a portion of these results. Many of the differentially expressed genes are known in other contexts to be involved in differentiation, motility, transcription, immunity, angiogenesis, extracellular matrix dissolution, or lipid metabolism. One sixth were nonannotated or encoded hypothetical proteins. Modeling based on structural homology revealed potential functions for 31 of these proteins. These data provide a reference set for understanding the molecular components of the dialogue taking place between maternal and fetal cells in the basal plate as well as for future comparisons of alterations in this region that occur in obstetric complications.


Asunto(s)
Perfilación de la Expresión Génica , Edad Gestacional , Intercambio Materno-Fetal , Placenta/metabolismo , Embarazo/metabolismo , Nacimiento a Término/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Modelos Biológicos
15.
Semin Perinatol ; 31(1): 10-8, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17317422

RESUMEN

Continued but slow progress has led to recent advances in our understanding that congenital cytomegalovirus (CMV) infection has occurred. We understand that the most severe congenital disease occurs following a primary maternal infection during pregnancy. We now have the ability to accurately diagnosis a primary maternal infection using serologic studies of single serum sample. For pregnant women with young children, we know that child-to-mother CMV transmission can probably be prevented by hygienic intervention, and that for pregnant women who have acquired a primary CMV infection, mother-to-fetal transmission is probably preventable using CMV hyperimmune globulin. Although additional studies are needed, treatment of congenitally infected fetuses or newborns should be possible using either CMV hyperimmune globulin or antiviral agents such as ganciclovir or its derivates. Finally, recent evidence indicates that CMV replicates in the placenta, impairs development, and causes inflammation and dysfunction. This plus the reversibility of many manifestations of congenital infection in the fetus and newborn indicate that congenital CMV disease is in part a syndrome of placental insufficiency.


Asunto(s)
Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/terapia , Enfermedades Fetales/terapia , Placenta/patología , Complicaciones Infecciosas del Embarazo/terapia , Infecciones por Citomegalovirus/prevención & control , Femenino , Enfermedades Fetales/prevención & control , Humanos , Recién Nacido , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/prevención & control , Factores de Riesgo
17.
Placenta ; 59 Suppl 1: S8-S16, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28477968

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is a major viral cause of birth defects, including microcephaly, neurological deficits, loss of hearing and vision, and intrauterine growth restriction. Despite its public health significance, there is no approved treatment for congenital infection during pregnancy; existing antivirals have unacceptable toxicities. The mechanisms of HCMV-induced placental injury, reduced capacity for compensatory development and transmission to the fetus are poorly understood, limiting the development of alternative strategies for clinical management of the disease. Recently, self-renewing, multipotent trophoblast progenitor cells (TBPCs) were reported to reside in the chorion of the human placenta and differentiate into the mature trophoblast subtypes - transport syncytiotrophoblasts and invasive cytotrophoblasts - forming chorionic villi, the functional units of the placenta. HCMV infects TBPCs, reducing the population of progenitor cells and their functional capacity to self-renew, migrate and differentiate. Human TBPCs and chorionic villus explants from first trimester represent relevant models for evaluating efficacies of new antiviral agents in protecting and restoring growth of the developing placenta in response to adverse conditions. Correlating pathology from complications of congenital HCMV infection with impaired development in the tissue environment of anchoring villus explants and defects in TBPC differentiation may enable identification of molecular pathways that could serve as targets for intervention. Here we summarize studies that could open up novel avenues of research on potential therapeutics to sustain placental development, promote differentiation and improve function and pregnancy outcomes.


Asunto(s)
Infecciones por Citomegalovirus/fisiopatología , Placentación , Complicaciones del Embarazo/fisiopatología , Anticuerpos Monoclonales/uso terapéutico , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/terapia , Femenino , Humanos , Embarazo , Trofoblastos/virología , Proteínas del Envoltorio Viral/inmunología
18.
Curr Opin Virol ; 27: 48-56, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29172071

RESUMEN

The emergence of congenital Zika virus (ZIKV) disease, with its devastating effects on the fetus, has prompted development of vaccines and examination of how ZIKV breaches the maternal-fetal barrier. Infection of placental and decidual tissue explants has demonstrated cell types at the uterine-placental interface susceptible to infection and suggests routes for transmission across the placenta and amniochorionic membrane. ZIKV replicates in proliferating Hofbauer cells within chorionic villi in placentas from severe congenital infection. Explants of anchoring villi recapitulate placental architecture and early-stage development and suggest infected Hofbauer cells disseminate virus to fetal blood vessels. ZIKV infection of explants represents a surrogate human model for evaluating protection against transmission by antibodies in vaccine recipients and passive immune formulations and novel therapeutics.


Asunto(s)
Placenta/virología , Primer Trimestre del Embarazo , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Animales , Bioensayo/métodos , Proliferación Celular , Femenino , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Técnicas de Cultivo de Órganos/métodos , Embarazo , Vacunas Virales , Cultivo de Virus/métodos , Virosis/virología , Replicación Viral , Virus Zika/fisiología , Infección por el Virus Zika/congénito , Infección por el Virus Zika/transmisión
19.
Lancet Infect Dis ; 17(7): e197-e208, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28433705

RESUMEN

Maternal immunisation has the potential to substantially reduce morbidity and mortality from infectious diseases after birth. The success of tetanus, influenza, and pertussis immunisation during pregnancy has led to consideration of additional maternal immunisation strategies to prevent group B streptococcus and respiratory syncytial virus infections, among others. However, many gaps in knowledge regarding the immunobiology of maternal immunisation prevent the optimal design and application of this successful public health intervention. Therefore, we did an innovative landscape analysis to identify research priorities. Key topics were delineated through review of the published literature, consultation with vaccine developers and regulatory agencies, and a collaborative workshop that gathered experts across several maternal immunisation initiatives-group B streptococcus, respiratory syncytial virus, pertussis, and influenza. Finally, a global online survey prioritised the identified knowledge gaps on the basis of expert opinion about their importance and relevance. Here we present the results of this worldwide landscape analysis and discuss the identified research gaps.


Asunto(s)
Inmunidad Materno-Adquirida , Inmunización/métodos , Complicaciones Infecciosas del Embarazo/prevención & control , Países en Desarrollo , Femenino , Salud Global , Humanos , Inmunización/estadística & datos numéricos , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/prevención & control , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Atención Prenatal/métodos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/administración & dosificación , Streptococcus agalactiae/aislamiento & purificación
20.
Trends Microbiol ; 13(4): 164-74, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15817386

RESUMEN

During human gestation, viruses can cause intrauterine infections associated with pregnancy complications and fetal abnormalities. The ability of viruses to spread from the infected mother to the fetus arises from the architecture of the placenta, which anchors the fetus to the uterus. Placental cytotrophoblasts differentiate, assume an endothelial phenotype, breach uterine blood vessels and form a hybrid vasculature that amplifies the maternal blood supply for fetal development. Human cytomegalovirus - the major cause of congenital disease - infects the uterine wall and the adjacent placenta, suggesting adaptation for pathogen survival in this microenvironment. Infection of villus explants and differentiating and/or invading cytotrophoblasts offers an in vitro model for studying viruses associated with prenatal infections.


Asunto(s)
Infecciones por Citomegalovirus/transmisión , Enfermedades Fetales/virología , Placenta/virología , Complicaciones Infecciosas del Embarazo/virología , Útero/virología , Virosis/transmisión , Animales , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/virología , Femenino , Humanos , Circulación Placentaria , Embarazo , Enfermedades Uterinas/virología , Virosis/congénito , Virosis/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA